Language selection

Search

Patent 2818890 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent: (11) CA 2818890
(54) English Title: METHOD AND SYSTEM FOR PROGNOSIS AND TREATMENT OF DISEASES USING PORTFOLIO OF GENES
(54) French Title: METHODE ET SYSTEME POUR LE PRONOSTIC ET LE TRAITEMENT DE MALADIES A L'AIDE D'UN PORTEFEUILLE DE GENES
Status: Granted
Bibliographic Data
(51) International Patent Classification (IPC):
  • C12Q 1/68 (2018.01)
  • C12Q 1/6809 (2018.01)
  • G16B 25/10 (2019.01)
  • G16B 50/00 (2019.01)
  • C40B 30/04 (2006.01)
(72) Inventors :
  • KANDULA, MAHESH (India)
(73) Owners :
  • KRISANI BIOSCIENCES (P) LTD (India)
(71) Applicants :
  • KRISANI BIOSCIENCES (P) LTD (India)
(74) Agent: MILTONS IP/P.I.
(74) Associate agent:
(45) Issued: 2022-07-12
(86) PCT Filing Date: 2011-06-21
(87) Open to Public Inspection: 2012-05-31
Examination requested: 2016-03-21
Availability of licence: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/IN2011/000417
(87) International Publication Number: WO2012/070056
(85) National Entry: 2013-05-23

(30) Application Priority Data:
Application No. Country/Territory Date
3524/CHE/2010 India 2010-11-23
61/429,857 United States of America 2011-01-05

Abstracts

English Abstract

This disclosure generally relates to a method, a system and a kit for diagnosing and treating various disease conditions using the gene expression value in a given sample obtained from a subject. This disclosure further relates to selecting the right set of genes as a portfolio of genes for a particular disease. Providing a set of appropriate portfolio of gene for studying the gene expression values and using the values as a diagnostic tool is also disclosed. The gene expression value is further used for intervention by pharmaceuticals as a prediction model for the treatment of the disease. An individual may have a personalized health card that may store the gene expression value for that particular individual. A system may store the data for portability and integrated view of the individual's medical data for effective diagnosis and treatment.


French Abstract

Cette invention concerne généralement une méthode, un système et une trousse pour le diagnostic et le traitement de divers états pathologiques à l'aide de la valeur d'expression génique dans un échantillon donné provenant d'un sujet. Cette invention concerne en outre la sélection du bon ensemble de gènes en tant que portefeuille de gènes pour une maladie particulière. L'invention concerne également l'apport d'un ensemble de portefeuilles appropriés de gènes pour l'étude des valeurs d'expressions géniques et l'utilisation des valeurs en tant qu'outil de diagnostic. La valeur d'expression génique est en outre utilisée pour une intervention par des produits pharmaceutiques en tant que modèle de prédiction pour le traitement de la maladie. Un individu peut avoir une carte de soin personnalisé qui peut stocker la valeur d'expression génique pour cet individu particulier. Un système peut stocker les données pour donner une portabilité et permettre une vision intégrée des données médicales de l'individu pour un diagnostic et un traitement efficaces.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
What is claimed is:
1. A method comprising: in a breast tissue sample from a subject suffering
from a disease,
measuring gene expression values for a portfolio of genes comprising BRCA1,
BRCA2, 1VIET,
PR, PI3KA, EGFR, PTEN, BRAF, kRAS, Her2 and ESR1 based on determining an
amount of
RNA produced by each gene that can code for one of: a protein and a peptide;
analyzing, through
a processor, the gene expression values for the portfolio of genes within said
sample through
repeatedly applying at least one of: penalized discriminant analyses and
recursive feature
elimination, said gene expression of said portfolio of genes being measured
using primer pairs:
SEQ ID NO.s 22 & 23, SEQ ID NO.s 24 & 25, SEQ ID NO.s 26 & 27, SEQ ID NO.s 28
& 29,
SEQ ID NO.s 30 & 31, SEQ ID NO.s 32 & 33, SEQ NO.s 34 & 35, SEQ NO.s 36 & 37,
SEQ ID NO.s 38 & 39, SEQ ID NO.s 40 & 41, and SEQ ID NO.s 42 & 43; obtaining,
through
the processor, a gene expression value profile of the portfolio of genes for
the sample from the
subject based on the analysis of the gene expression values, the gene
expression value profile
comprising a statistically significant number of genes that vary in expression
value from an
expression value of the same genes in at least one of: a normal and a disease
control sample; and
normalizing, through the processor, a gene expression level of the portfolio
of genes using a
house keeping gene, the at least one of: the normal and the disease control
sample and the sample
from the subject in addition to obtaining the gene expression value profile,
the house keeping
gene being GAPDH.
2. The method of claim 1, further comprising: performing a statistical
analysis on the gene
expression level for the portfolio of genes, a statistically significant value
associated therewith
representing collectively a level of gene expression values for the portfolio
of genes that
indicates that said subject has at least one of: a cancer, a progression of
the cancer, an effect of
treatment for the cancer and a location of the cancer.
3. The method of claim 1, wherein the sample is at least one of: a biopsy
sample, a fine needle
aspirate, parafilm embedded samples, a tumor tissue, immune cells, whole
blood, saliva, urine,
3 7
Date Recue/Date Received 2021-04-09

synovial fluid, bone marrow, cerebrospinal fluid, vaginal mucus, cervical
mucus, nasal
secretions, sputum, semen, amniotic fluid, bronchoalveolar lavage fluid, and
other cellular
exudates from a patient having cancer.
38
Date recue / Date received 2021-11-02

Description

Note: Descriptions are shown in the official language in which they were submitted.


WO 2012/070056
PCT/1N2011/000,417
METHOD AND SYSTEM FOR PROGNOSIS AND TREATMENT OF DISEASES
USING PORTFOLIO OF GENES
CROSS REFERENCE TO RELATED APPLICATION
[001] This disclosure claims priority to pending US Provisional application
61/429857
filed on 5th January 2011 and Indian Provisional application 3524/CHE/2010
filed on
23/11/2010.
FIELD OF TECHNOLOGY
[002] This disclosure generally relates to a method, a system and a kit for
diagnosing
and treating various disease conditions using the gene portfolio in a subject.
More
specifically, the portfolio of gene and their expression value may be used for
tool by the
treatment provider as a prognostic model and change in treatment regimen for a
particular
disease.
BACKGROUND
[003] In women, two of the most widely used screening tests are the
Papanicolaou
(PAP) test to detect cervical cancer and mammography to detect breast cancer.
Both
screening tests have been successful in reducing the death rates from these
cancer in
certain age groups. There are still many cases that are not detected through
these .
techniques and the mortality rate is very high due to high cost, lack of
insurance coverage
and reluctance.
[004] In the case of breast cancer, great efforts have been made to develop a
detection
test by mammography. Although several studies indicate that mass mammography
may
be a useful strategy to reduce breast cancer mortality, this method involves a
certain
number of disadvantages. Some of the disadvantages are a high rate or false
positives,
frequent false negatives and enormous public health costs. Thus, when the
benefits are
weighed against these advantages, it is not surprising that this form of
screening has
engendered contentious debates over the last twenty 30 years.
[005] In men, prostate-specific antigen (PSA) levels in the blood may be used
to screen
for prostate cancer. PSA levels are high in men with prostate cancer, but
levels also are
elevated in men with noncancerous (benign) enlargement of the prostate.
Currently the
-1 -
CA 2818890 2019-07-04

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
main drawback to its use as a screening test is the large number of false-
positive results,
which generally lead to more invasive tests.
[006] It is known that diagnosis and follow up of the evolution of cancer are
carried out,
besides direct observation of the tumors, by biopsy analysis or in the case of
blood
malignancies by analysis of the bone marrow, which implies either a surgical
intervention, or an invasive test such as a biopsy or a bone marrow
aspiration. Now, in
addition to the disagreeable or even dangerous aspect of such methods, it has
been
observed that they may not be very precise. Current methods for classifying
human
malignancies are mostly to rely on a variety of morphological, clinical and
molecular
variables. Despite recent progress, there are still many uncertainties in
diagnosis.
Furthermore, it is likely that the existing classes of the tumors are
heterogeneous and
comprise diseases that are molecularly distant.
SUMMARY
[007] In this disclosure a method, system and kit are being presented which
may be used
for diagnosis, prognosis and/or for monitoring the treatment regimen. In one
embodiment,
a method of diagnosing a disease using a portfolio of genes that is specific
for an
individual or for the disease or for a subject is being disclosed. In another
embodiment,
gene expression values are measured and used for evaluating the presence
and/or absence
of the disease condition in a subject.
[008] In another embodiment, a method of developing a gene expression values
for the
portfolio of genes that are specific to the profile indicative of the presence
or stage of a
selected disease, disorder or genetic pathology is being presented.
[009] In one embodiment, a mathematical algorithm is executed on a computer
readable
medium to select a portfolio of genes to diagnose a particular disease using
the metabolic
pathway.
[010] In another embodiment, a predictive mathematical expression value is
displayed
to show the portfolio of genes affected for a specific disease. In another
embodiment,
suggestive inhibitors and enhancers for a particular gene expression value are
proposed as
a pharmaceutical treatment option. In another embodiment, a treatment provider
may
input the existing treatment regime and observe the effect of the treatment
over a period
of time using the gene expression value.
-2-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
[011] In another embodiment, a system that is integrated for patient
treatment, analysis,
diagnosis and prognosis are provided for the treatment. In another embodiment,
a
personalized medical card may be used (with data storage capacity) for each
specific
patient (subject) that can provide his or her disease diagnosis gene
expression value for a
portfolio of genes, treatment pattern and analysis. The data generated will be
stored in
each card as well as updated to the central database server. The information
of each
patient will be stored and shared with the insurance company, health care
personal and
institution that provide health care.
[012] In another embodiment, the gene expression value is analyzed using a
penalized
discriminate analysis with recursive feature elimination. This disclosure
particularly
describes cancer diagnosis and treatment for an individual.
[013] A method of diagnosing a cancer in a mammalian subject includes the
steps of
examining a sample containing the subject's samples and detecting a variance
in the gene
expression values for the portfolio of genes that are statistically
significant in number,
e.g., at least 10 (not limited to) tumor and non-tumor genes from those same
genes in a
characteristic disease or healthy gene expression value. A significant
variance in the gene
expression value of these genes when compared to an average gene expression
value
profile of a normal control, or significant similarities to an average gene
profile of
subjects with cancer, correlates with a specific type of cancer and/or the
location of
tumor. The present disclosure relates to a method of diagnosis and/or follow
up of several
types of cancer, for instance after a chemotherapy or after an operation.
[014] In one embodiment, the method of diagnosis and/or follow up of the
evolution of
cancer includes the analysis of the RNA component and the mRNA coding for the
proteins of the ribonucleoprotein telomerase in the blood plasma or serum.
[015] The present disclosure accommodates the use of tissue and blood for gene

expression value analysis but not limited to archived paraffin-embedded biopsy
material,
aspirates, fine needle aspirates, and any biological sample from mammalian
subject for
assay of all marker in the set. It is also compatible with several different
methods of
tumor tissue harvest, for example, via core biopsy or fine needle aspiration.
Further, for
each member of the portfolio of genes, the disclosure specifies
oligonucleotide sequences
that can be used in the test.
-3-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
[016] In one embodiment, a method of diagnosing cancer is performed by
identifying
differential modulation of each gene (relative to the expression value of the
same genes in
a normal population) in a combination of portfolio of genes comprising of
pathology
associated pathways.
[017] Accordingly, in one embodiment a linear sequence of a polynucleotide
comprising
a selected from the portfolio of genes (10-45 nucleotides long) comprises of
TRP channel
encoding genes, Breast cancer encoding genes such as HER-2 and its subtypes.
Preferably, said RNA is overabundant in a proportion of breast cancer cells.
In one
embodiment, of this disclosure is an isolated polynucleotide comprising a
linear sequence
is represented by SEQ. ID NO: 01 to SEQ. ID NO: 061. These embodiments include
an
isolated polynucleotide which is a DNA polynucleotide, an RNA polynucleotide,
a
polynucleotide probe, or a polynucleotide primer.
[018] In one embodiment, the RNA is overabundant by at least about 20% of a
representative panel of breast cancer cell lines; more preferably, it is
overabundant by at
least about 40% of the panel; even more preferably, it is overabundant by at
least 60% or
more. In a further embodiment, RNA is isolated from a fixed, wax-embedded
breast
cancer tissue specimen of the patient.
[019] In a different aspect, the disclosure concerns a method of preparing a
personalized
genomics profile for a patient, comprising the steps of:
(a) subjecting RNA extracted from a breast tissue obtained from the patient to
gene
expression analysis; (b) determining the expression level of one or more genes
selected
from the breast cancer genes, wherein the expression level is normalized
against a control
gene or genes and optionally is compared to the amount found in a breast
cancer
reference tissue set; and (c) creating a report summarizing the data obtained
by the gene
expression analysis. (d) storing the data and sharing it with the stake
holders such as
physicians, insurance companies and hospitals.
[020] In one embodiment, a method of diagnosing breast cancer cells is
disclosed. Still
another embodiment of this disclosure is a diagnostic kit for detecting or
measuring
specific gene expression value present in clinical samples; comprising a
reagent, and a
buffer in suitable packaging, wherein the reagent comprises of the RNAse
inhibitor,
TRIZOLTm reagent, ethanol, dNTPs, Reverse Transcriptase enzyme, stabilizing
buffer
-4-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
solutions, DNA polymerases, SYBER green dNTPs, target polynucleotide's as
primers
for gene expression value analysis and magnesium chloride.
[021] The present disclosure relates to the diagnosis of cancer, and more
specifically to
the identifying and measuring the portfolio of gene expressed by circulating
cells of the
immune system and/or circulating cancer cells and/or tumor cells. In one
embodiment,
identification of differentially modulated gene (relative to the expression
value of the
same genes in a normal population) is performed using a combination of genes
selected
from the group comprising of SEQ ID NO: 001 to 061.
[022] Yet another embodiment, the gene expression value of the immune cell
from the
peripheral blood is used as a diagnostic parameter to predict the disease
state.
BRIEF DESCRIPTION OF DRAWINGS
[023] Figure 1 shows a method and treatment using gene expression value 100 to
treat a
subject having cancer.
[024] Figure 2 shows a flow chart of analysis of breast cancer relative gene
expression
200.
[025] Figure 3 shows real time PCR program overview 300 for one of the RT-PCR
reaction.
[026] Figure 4 shows proposed treatment 400 blocks for certain genes that are
over
expressed and the suitable inhibitors.
DETAILED DESCRIPTION
[027] The instant disclosure describes a method, a system and a kit for
diagnosing and
treating various disease conditions using the gene expression value in an
individual. The
present disclosure describes a novel method for the diagnosis of cancer based
on gene
expression value for a given portfolio of genes.
[028] Figure 1, shows an individual 110 having a disease condition that is
being
diagnosed 120 using the gene expression value after the physician or the
medical care
personal has accessed the background and performed routine clinical tests.
Suitable
treatment 130 is performed using the conventional method or proposed treatment
400.
The medical data thus produced is stored in a personalized card 180. A system
comprising of several client servers 140A..N is used for accessing, storing,
analyzing and
prescribing the medical treatment for the individual 110. The data is stored
in database
- -5-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
160 for future use. The current system is portable and may be used for
electronic medical
record keeping and updating other information such as personal identification
number,
medical record number, physical check up data etc.
[029] Figure 2, shows a flow chart for the treatment method. The treatment
method may
involve the analysis of an array of gene expression value for a portfolio of
genes from a
biological sample of one or more subjects having the disease in question by
applying
penalized discriminant analysis and recursive feature elimination steps to the
gene
expression value.
[030] The recursive feature elimination identifies and eliminates the least
informative
up-regulated and down-regulated genes from the first profile. These steps are
optionally
repeated until a gene expression value profile is obtained containing
statistically
significant number of genes that vary in expression value from the expression
value of the
same genes in the array of the healthy or disease control. This gene
expression value
profile is a characteristic of the selected disease, disorder or genetic
pathology or a stage
of the selected disease, disorder or genetic pathology. The profile of
variance in
expression value of the genes compared to a normal control or to a disease
control
correlates with the type and/or location of the disease, disorder or genetic
pathology.
[031] In one embodiment, the gene expression value profiles of the immune
cells
circulating in the peripheral blood of cancer patients reflect the presence of
a solid tumor.
Preferably, for cancer characterized by a solid tumor, the genes examined for
the profiles
are genes normally expressed by the patients' immune cells. For cancer in
which the
tumor or cancer cells circulate in the peripheral blood, e.g., CTCL, is a
specific example
of cancer or tumor now present in circulating in blood.
[032] Each gene is sufficiently specific to indicate the type and location of
the cancer or
tumor. In some embodiments, this characteristic gene expression value is
detectable
earlier than any other sign of tumor presence. Gene expression value for
portfolio of
genes to be detected in peripheral blood samples thus is a powerful tool for
cancer
diagnosis and staging, as well as the monitoring of therapeutic efficacy.
[033] In one embodiment, for establishing gene expression value for portfolio
of gene
profile includes determining the amount of RNA that is produced by a gene that
can code
for a protein or peptide. This is accomplished by using methods such as
reverse
-6-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
transcriptase PCR (RT-PCR), competitive RT-PCR, real time RT-PCR, differential

display RT-PCR, Northern Blot analysis and other related tests.
[034] In one embodiment, a portfolio of genes may be designed in such a way
that they
are clinically relevant for making a diagnosis, prognosis, or treatment
choice. These sets
of genes make up the portfolio of genes for the disclosure.
[035] Genes that display similar expression value patterns may be co-regulated
by an
identical factor that regulates the genes in the same direction. Choosing an
optimal
portfolio of genes is beneficial for providing a sensitive and accurate
diagnostic kit.
[036] In the method of the disclosure, a group of genetic marker is selected
for use in
diagnostic applications. These groups of marker are "portfolios". Diagnostic
applications
include the detection or identification of a disease state or condition of a
subject,
determining the likelihood that a subject will may suffer from a disease or
condition,
determining the likelihood that a subject with a disease or condition will
respond to
therapy, determining the prognosis of a subject with a disease or condition
(or its likely
progression or regression), and determining the effect of a treatment on a
subject with a
disease or condition. For example, the method can be used to establish
portfolios for
detecting the presence or likelihood of a subject contracting colon cancer or
the likelihood
that such a subject will respond favorably to cytotoxic drugs.
[037] The portfolio of gene selected by the method of the disclosure contain a
number
and type of marker that assure accurate and precise results and are economized
in terms
of the number of genes that comprise the portfolio.
[038] Most preferably, the markers employed in the portfolio are nucleic acid
DNA
Sequences that express mRNA ("genes"). Expression value of the marker may
occur
ordinarily in a healthy subject and be more highly expressed or less highly
expressed
when an event that is the object of the diagnostic application occurs.
Alternatively,
expression value may not occur except when the event that is the object of the
diagnostic
application occurs.
[039] Distinctions are made among the diagnostic parameters through the use of

mathematical/statistical values that are related to each other. The preferred
distinctions
are mean signal readings indicative of gene expression value and measurements
of the
variance of such readings.
-7-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
[040] A relationship between each genes baseline and experimental value must
first be
established. The preferred process is conducted as follows. A baseline class
is selected.
Typically, this will comprise of genes from a population that does not have
the condition
of interest. For example, if one were interested in selecting a portfolio of
genes that are
diagnostic for breast cancer, samples from patients without breast cancer can
be used to
make the baseline class. Once the baseline class is selected, the arithmetic
mean and
standard deviation is calculated for the indicator of gene expression value of
each gene
for baseline class samples.
DEFINITIONS
[041] "Patient" or "subject" as used herein means a mammalian animal,
including a
human, a veterinary or farm animal, a domestic animal or pet, and animals
normally used
for clinical research.
[042] "Sample" as used herein means any biological fluid or tissue that
contains immune
cells and/or cancer cells. A suitable sample for use in this disclosure,
whether the cancer
is a solid tumor cancer or a cancer characterized by circulating cancer cells,
includes
peripheral blood. Other useful biological samples include, without limitation,
fine needle
aspirants, parafilm embedded tissue, whole blood, saliva, urine, synovial
fluid, bone
marrow, cerebrospinal fluid, vaginal mucus, cervical mucus, nasal secretions,
sputum,
semen, amniotic fluid, bronchoalveolar lavage fluid, and other cellular
exudates from a
patient having cancer. Such samples may further be diluted with saline, buffer
or
physiologically acceptable diluents. Alternatively, such samples are
concentrated by
conventional means.
[043] "Immune cells" as used herein means B-lymphocytes, T-Iymphocytcs, NK
cells,
macrophages, mast cells, monocytes and dendritic cells.
[044] The term "polynucleotide," when used in singular or plural, generally
refers to any
polyribonucleotide or poly deoxyribonucleotide, which may be unmodified RNA or
DNA
or modified RNA or DNA. Thus, for instance, polynucleotides as defined herein
include,
without limitation, single- and double-stranded DNA, DNA including single and
double-
stranded regions, single- and double-stranded RNA, and RNA including single-
and
double-stranded regions, hybrid molecules comprising DNA and RNA that may be
single-stranded or, more typically, double-stranded or include single- and
double-
stranded regions. In addition, the term "polynucleotide" as used herein refers
to triple-
-8-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
stranded regions comprising RNA or DNA or both RNA and DNA. The strands in
such
regions may be from the same molecule or from different molecules. The regions
may
include all of one or more of the molecules, but more typically involve only a
region of
some of the molecules. One of the molecules of a triple-helical region often
is an
oligonucleotide. The term "polynucleotide" specifically includes cDNAs. The
term
includes DNAs (including cDNAs) and RNAs that contain one or more modified
bases.
Thus, DNAs or RNAs with backbones modified for stability or for other reasons
are
"polynucleotides" as that term is intended herein. Moreover, DNAs or RNAs
comprising
unusual bases, such as inosine, or modified bases, such as tritiated bases,
are included
within the term "polynucleotides" as defined herein. In general, the term
"polynucleotide"
embraces all chemically, enzymatically and/or metabolically modified forms of
unmodified polynucleotides, as well as the chemical forms of DNA and RNA
characteristic of viruses and cells, including simple and complex cells.
[045] The term "oligonucleotide" refers to a relatively short polynucleotide,
including,
without limitation, singlestranded deoxyribonucleotides, single- or double-
stranded
ribonucleotides, RNA:DNA hybrids and double-stranded DNAs. Oligonucleotides,
such
as single-stranded DNA probe oligonucleotides, are often synthesized by
chemical
methods, for example using automated oligonucleotide synthesizers that are
commercially available. However, oligonucleotides can be made by a variety of
other
methods, including in vitro recombinant DNA -mediated techniques and by
expression of
DNAs in cells and organisms.
[046] The terms "differentially expressed gene," "differential gene
expression" and their
synonyms, which are used interchangeably, refer to a gene whose expression is
activated
to a higher or lower level in a subject suffering from a disease, specifically
cancer, such
as breast cancer, relative to its expression in a normal or control subject.
The terms also
include genes whose expression is activated to a higher or lower level at
different stages
of the same disease. It is also understood that a differentially expressed
gene may be
either activated or inhibited at the nucleic acid level or protein level, or
may be subject to
alternative splicing to result in a different polypeptide product. Such
differences may be
evidenced by a change in mRNA levels, surface expression, secretion or other
partitioning of a polypeptide, for example. Differential gene expression may
include a
comparison of expression between two or more genes or their gene products, or
a
comparison of the ratios of the expression between two or more genes or their
gene
-9-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
products, or even a comparison of two differently processed products of the
same gene,
which differ between normal subjects and subjects suffering from a disease,
specifically
cancer, or between various stages of the same disease. Differential expression
includes
both quantitative, as well as qualitative, differences in the temporal or
cellular expression
pattern in a gene or its expression products among, for example, normal and
diseased
cells, or among cells which have undergone different disease events or disease
stages. For
the purpose of this disclosure, "differential gene expression" is considered
to be present
when there is at least an about two-fold, preferably at least about four-fold,
more
preferably at least about six-fold, most preferably at least about ten-fold
difference
between the expression of a given gene in normal and diseased subjects, or in
various
stages of disease development in a diseased subject.
[047] As used herein, the term "cancer" means any cancer. In one embodiment,
the
cancer is characterized by the presence of a solid tumor. Among such cancer
are included,
without limitation, breast cancer, neuronal cancer, prostate cancer,
pancreatic cancer,
brain cancer, melanoma, other skin cancer, esophageal cancer, colorectal
cancer, ovarian
cancer, small cell carcinoma, adrenal cancer, lung adenocarcinoma,
mesothelioma,
Hodgkins lymphoma and non-Hodgkins Lymphoma without blood involvement. In
another embodiment, the cancer is characterized by the presence of circulating
cancer
cells in the peripheral blood, e.g., CTCL, T-ALL, B-ALL, CML, CLL, APL, AML, B-

CLL, or hairy cell leukemia. Suitable cancer for diagnosis or screening with
the methods
described below include early stage cancer or late stage cancer.
[048] By "non-tumor genes" as used herein is meant genes which are normally
expressed in other cells, preferably immune cells, of a healthy mammal, and
which are
not specifically products of tumor cells.
[049] -The term "statistically significant number of genes" in the context of
this
disclosure differs depending on the degree of change in gene expression value
observed.
The degree of change in gene expression value varies with the type of cancer
and with the
size or spread of the cancer or solid tumor. The degree of change also varies
with the
immune response of the individual and is subject to variation with each
individual. For
example, in one embodiment of this disclosure, a large change, e.g., 2-3 fold
increase or
decrease in a small number of genes, e.g., in from 5 to 8 characteristic
genes, is
statistically significant. In another embodiment, a smaller relative change in
about 30 or
more genes is statistically significant. This is particularly true for cancer
with solid
-10-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
tumors. Still alternatively, if a single gene is profiled as up-regulated or
expressed
significantly in cells which normally do not express the gene such up-
regulation of a
single gene may alone be statistically significant. Conversely, if a single
gene is profiled
as down regulated or not expressed significantly in cells which normally do
express the
gene, such down-regulation of a single gene may alone be statistically
significant.
[050] As an example, a single gene, which is expressed about the same in all
members
of a population of patients, is 4- fold down regulated in only 1% of
individuals without
cancer. Four such independently regulated genes in one individual, all 4 fold
down
regulated, would occur by chance only one time in 100 million. Therefore those
4 genes
are a statistically significant number of genes for that cancer.
Alternatively, if normal
variance is higher, e.g., one healthy person in 10 has the gene 4-fold down-
regulated, and
then a larger panel of genes is required to detect variance for a particular
cancer.
[051] Thus, the methods of this disclosure contemplate examination of the
expression
value profile of a "statistically significant number of genes" ranging from 1
to 100 genes
in a single profile. In one embodiment, the gene profile is formed by a
statistically
significant number of at least one gene. In another embodiment, the gene
profile is
formed by a statistically significant number of at least 4 genes. In still
another
embodiment' the gene profile is formed by at least 10 genes. In still other
embodiments,
the gene profiles examined as part of these methods, particularly in cases in
which the
cancer are characterized by solid tumors, contain, as statistically
significant numbers of
genes, 20, 30, 40, 50, 60, 70, 80, or 90 genes in a panel.
[052] The following table shows an optimized portfolio of genes for Leukemia.
Table 1:
Acute lymphoblastic - Acute myeloid Chronic lymphocytic
Chronic myeloid
leukemia (ALL) leukemia (AML) leukemia (CLL) leukemia (CML)
(alteration in the
(alteration in the gene/gene (alteration in the (alteration in the
gene/gene expression
expression value) gene/gene gene/gene expression value)
expression value)
value)
(precursor B lymphoblastic FLT3 (mutation) Bc1-2 BCR-ABL
leukemia) c-KIT (mutation) (overexpression (translocation)
N-ras (mutation) value) EVIl
BCR-ABL (translocation) K-ras (mutation) p53
(mutation) (overexpression
MLL (rearrangement] PML-RAR alpha ATM (germline and value)
-11-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
E2A-PBX1 (translocation) (translocation) somatic mutation)
AML1
c-MYC (translocation) AML 1 -ETO Fas (absent) (translocation)
TEL-AML1 (translocation) (translocation) JAK2(translocation)
PLZF-RAR
(precursor T lymphoblastic alpha p I 6/INK4A
leukemia) (translocation) (mutation)
AML I p53 (mutation)
NOTCH1 (mutation) (mutation) RB1 (mutation)
TAL1 (overexpression C/EBPalpha
value) (mutation)
LYL1 (expression value) PU.1 (mutation)
MLL-ENL (translocation)
HOX11 (translocation) CBFbeta-
MYC (translocation) MYH1 1
LMO2 (translocation) (inversion)
HOX11L2 (translocation)
[053] An important aspect of the present disclosure is to use the measured
expression of
certain genes by breast cancer tissue to provide prognostic information. For
this purpose it
is necessary to correct for (normalize away) both differences in the amount of
RNA
assayed and variability in the quality of the RNA used. Therefore, the assay
typically
measures and incorporates the expression of certain normalizing genes,
including well
known housekeeping genes, such as GAPDH and Cyp 1. Alternatively,
normalization can
be based on the mean or median signal (Ct) of all of the assayed genes or a
large subset
thereof (global normalization approach). On a gene-by-gene basis, measured
normalized
amount of a patient tumor mRNA is compared to the amount found in a breast
cancer
tissue reference set. The number (N) of breast cancer tissues in this
reference set should
be sufficiently high to ensure that different reference sets (as a whole)
behave essentially
the same way. If this condition is met, the identity of the individual breast
cancer tissues
present in a particular set will have no significant impact on the relative
amounts of the
genes assayed. Usually, the breast cancer tissue reference set consists of at
least about 30,
preferably at least about 40 different FPE breast cancer tissue specimens.
Unless noted
otherwise, normalized expression levels for each in RNA tested tumor/patient
will be
expressed as a percentage of the expression level measured in the reference
set. More
specifically, the reference set of a sufficiently high number (e.g. 40) of
tumors yields a
distribution of normalized levels of each mRNA species.
[054] The following is the portfolio of genes selected for Breast Cancer
detection,
prognosis or treatment.
-12-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
[055] BCR-ABL: NM 004327.3 and ABL1: NM 005157.3
Table 2: Primers used for analysis for the above mentioned genes:
Seq.ID Unique Gene ID DNA Sequence
001 BCRI GCTTCTCCCTGACATCCGTG
002 Al3L1 GGCCCATGGTACCAGGAGTG
003 BCR 2 GGAGCTGCAGATGCTGACCAAC
004 ABL2 G rri CTCCAGACTGTTGACTG
005 BCR3 CGCATGTTCCGGGACAAAAGC
006 BCR 4 CGCTCTCCCTCGCAGAACTC
007 BCRc30 AGAGGTCCAAGGTGCCCTAC
008 BCRc31 CGGACATCCAGGCACTGAAG
C30 and 31 refers to the codon position of the corresponding gene
[056] FLT3:NM_004119.2
Table 3: Primers used for analysis for the above mentioned gene:
Seq.ID Unique Gene ID DNA Sequence
009 FLT ITD F GCAATTTAGGTATGAAAGCCAGC
010 FLT ITD R C1TrCAGCAIITIGACGGCAACC
011 FLT D F CCGCCAGGAACGTGCTTG
012 FLT D R GCAGCCTCACATTGCCCC
[057j PML: NM 002675.3 and RARA: NM 000964.3
13

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Table 4: Primers used for analysis for the above mentioned genes:
Seq.ID Unique Gene ID DNA Sequence
013 PML ACCGATGGCTTCGACGAGTTC
014 RARA AGCCCTTGCAGCCCTCACAG
[058] JAK2:NM_004972.3
Table 5: Primers used for analysis for the above mentioned gene:
Seq.1D Unique Gene ID DNA Sequence
015 JK Forward (SP) AGCA ITIGGIT11 AAATTATGGAGTATATT
016 JK Forward (IC) ATCTATAGTCATGCTGAAAGTAGGAGAAAG
017 JK Reverse CTGAATAGTCCTACAGTG FIT1CAG Fri CA
[059] P53:NM_000546.4
Table 6: Primers used for analysis for the above mentioned genes are:
Seq.ID Unique Gene ID DNA Sequence
018 TP53FW GGCCCACTTCACCGTACTAA
019 TP53RE GTGGTTTCAAGGCCAGATGT
[060] EVI1, NM 001105077.3
Table 7: Primers used for analysis for the above mentioned gene:
S.No. Unique Gene ID DNA Sequence
020 EVI1 F CAAGGAAACTGGCCACAAAT
021 EVI1 R GGGGCT1TGTAAGGAGAACC
[061] A set of portfolio of genes optimized for breast cancer detection are as
follows:
BRCA1, BRCA2, MET, PR, PIK3A, EGFR, PTEN, BRAF, kRAS, Her2, ESR1.
-14-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
[062] BRCA1 : NM 007295.2
Table 8: Primers used for analysis for the above mentioned gene:
Seq.
ID Unique Gene ID DNA Sequence
022 BRCAlFw TGCTTGAAGTCTCCCTTG
023 BRCA1Re CTTCCATTGAAGGGTCTG
[063] BRCA2: NM 138081.2
Table 9: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
024 BRCA2Fw ACCC r1-1 CAGGTCTAAATGG
025 BRCA2Re TGCCTGCTTTACTGCAAG
[064] MET: NM_078571.2
Table 10: Primers used for analysis for the above mentioned gene:
Seq.ID Unique Gene ID DNA Sequence
026 MET Fw GAAGACCTTCAGAAGGTTG
027 MET Re TGGGGAGAATATGCAGTG
[065] PR: NM_001172044.1
Table 11: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
028 PR-Fw GGACTTGTGAGTACTCTG
029 PR-Re AGTGGGTGTTGAATGTG
[066] P131(A:_NM_006218.2
-15-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Table 12: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
030 PI3KA Fw GGACAATCGCCAATTCAG
031 PI3KA Re TGGTGGTGCTITI GATCTG
[067] EGFR: NM_057410.3
Table 13: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
032 EGFR Fw TTAGCAGGAAAGGCACTG
033 EGFR Re CAGCTTCATCCTACACAAG
[068] PTEN: NM 058074.3
Table 14: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
034 PTEN Fw GACAGACTGATGTGTATACG
035 PTEN Re GTGTAAATAGCTGGAGATGG
[069] BRAF: NM 139294.5
Table 15: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
036 BRAF Fw ATGGTGATGTGGCAGTGAAA
037 BRAF Re TAGCCAGTTGTGGCTTTGTG
[070] kRAS: NM 021284.6
-16-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Table 16: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
038 kRAS Fw AGGGCAGTTTGGATAGCTCA
039 WAS Re CACCACCCCAAAATCTCAAC
[071] Her2: NM_131089.1
Table 17: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
040 Her2Fw AGTACCTGGGTCTGGACGTG
041 Her2Re CTGGGAACTCAAGCAGGAAG
[072] ESR1: M14_012689.1
Table 18: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
042 ESR1Fw CTTGTGCAGGATTGTTGTG
043 ESR1Re GCCAATTGTAGGAACACAG
[073] The following is the portfolio of genes selected are for brain cancer
detection:
[074] TW, EMP3, OLIG2, IGFBP2, LGALS3, AQP1, TP53, EVI1, IL3
[075] TW :NM_022093.1
Table 19: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
044 TW Fw CTGTCCTGGGCAGAAAGAAG
045 TW Re GGTGAGAGGGAAGGAACCTC
[076] EMP3: NM 001425.2
17

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Table 20: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
046 EMP3 Fw GAGAGCGAGCGAGAGAGAAA
047 EMP3 Re GCTGGAGTCGGAGTCTTGTC
[077] OLIG2 :NM_005806.2
Table 21: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
048 OLIG2Fw CAGAAGCGCTGATGGTCATA
049 OLIG2Re TCGGCAGTTTTGGGTTATTC
[078] IGFBP2 : NM 000597.2
Table 22: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
050 IGFBP2 Fw GCAGAAAACGGAGAGTGCTT
051 IGFBP2 Re AAAGCAAGAAGGAGCAGGTG
[079] LGALS3: NM 002306.3
Table 23: Primers used for analysis for the above mentioned gene:
Unique Gene
Seq. ID ID DNA Sequence
052 LGALS3 Fw GGCCACTGATTGTGCCTTAT
053 LGALS3 Re TCTTTCTTCCCTTCCCCAGT
[080] AQP1 : NM_001185060.1
- -18-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Table 24: Primers used for analysis for the above mentioned gene:
Seq.ID Unique Gene ID DNA Sequence
054 AQP1 Fw ATTAACCCTGCTCGGTCCTT
055 AQP1 Re ACCCTGGAGTTGATGTCGTC
[081] TP53 :NM_030989.3
Table 25: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
056 P53 Fw GGCCCACTTCACCGTACTAA
057 P53 Re GTGGTTTCAAGGCCAGATGT
[082] EV11: NM 001105077.3
Table 26: Primers used for analysis for the above mentioned gene:
Unique Gene
Seq. ID ID DNA Sequence
058 EVIl Fw CAAGGAAACTGGCCACAAAT
059 EVIl Re GGGGCTTTGTAAGGAGAACC
[083] IL3 : NM_000588.3
Table 27: Primers used for analysis for the above mentioned gene:
Seq. ID Unique Gene ID DNA Sequence
060 113 Fw CTTTGCCTTTGCTGGACTTC
061 113 Re CCGTCCTTGATATGGATTGG
[084] After the forward (Fw) and reverse (Re) primer design for each portfolio
of genes
selected the next step is to extract total RNA extraction and the following
protocol was
used. Mix 0.75 ml of TRI Reagent RT - Blood with 0.25 ml of whole blood,
plasma or
-19-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
serum. Close the tube and shake the resulting lysate by hand or vortex.
Supplement the
homogenate with 50 I of chloroform per 0.75 ml of TRI Reagent RT - Blood used
for
lysis. Cover the samples tightly and shake vigorously for 15 seconds.
Centrifuge the
resulting mixture at 12,000 g for 15 minutes at 4 C. Following centrifugation,
the mixture
separates into a lower, red phenol phase, the interphase, and the upper
aqueous phase.
RNA remains in the aqueous phase whereas DNA and proteins are in the
interphase and
organic phase. Transfer 0.5 ml of the aqueous phase to a fresh tube.
Precipitate RNA from
the aqueous phase by mixing it with 0.5 ml of isopropanol. Store samples at
room
temperature for 5 - 10 minutes and centrifuge at 4,000 - 12,000 g for 5
minutes at 4 - 25
C. Remove the supernatant and wash the RNA pellet with 1 ml of 75% ethanol by
vortexing and subsequent centrifugation at 6,000 g for 5 minutes at 4 -25 C.
Remove the
ethanol wash and add water to the RNA pellet. Avoid drying the pellet as this
will
decrease its solubility.
[085] Once the total RNA is extracted the first step is to perform cDNA
amplification,
which is done as follows:
[086] Step: 1: Reaction mix I for cDNA amplification.
Step Total RNA (lng ¨ lug) 1-10 1
1 Random primer (50pm4t1) 2 1
2 dNTPs (10mM) 4 111
3 Total volume 16 I
4 Incubate the reaction at 70 C for 5min
[087] Add the following to the reaction mix II into Reaction mixlto perform:
-20-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
[088] Step: 2 Reaction mix II for cDNA amplification
lx Reaction buffer 2 1
100U/ ttl RNAse inhibitor 1 pl
1 unit of Reverse transcriptase 1 I
Total volume 4. I
[089] Mix Reaction I and II well by pipetting and setup the reverse
transcription
reaction and perform Step 3.
[090] Step: 3: Thermal cycling conditions for cDNA amplification (reverse
transcription)
Cycle No Temperature and time
1 25 C forl 0 min
2 42 C for 60 min
3 95 C for 5 min
[091] Real time PCR analysis: Add appropriate gene specific primers with
prepared
cDNA and subject to RT-PCR. Real time PCR mix contains cDNA 1 1, Forward
primer
(10pm/ 1) 0.4 p1, Reverse Primer (10pm/ 1) 0.4 pl, EvaGreen Mix (2X), 12.5
I, H20
5.7 I, and the total volume would be 20 1.
[092] Real time PCR condition, also shown in Figure 3. The cycle number 3 is X
C and
it depends on the primer being used and their respective annealing
temperature.
[093] Portfolio of Genes, their amplified product and their respective
annealing
temperature.
-21-

CA 02818890 2013-05-23
WO 2012/070056
PCT/1N2011/000417
Table 28:
Amplified
Marker Ann
Forward Primer Reverse Primer Product
Name Temp
Size
BRCA2 ACCCTTTCAGGTCTAAATGG TGCCTGCTTTACTGCAAG 268
54
MET GAAGACCTTCAGAAGGTTG TGGGGAGAATATGCAGTG 267
PR GGACTTGTGAGTACTCTG AGTGGGTGTTGAATGTG 266
57
PIK3A
GGACAATCGCCAATTCAG TGGTGGTGC1-11 GATCTG 268
BRCA1 TGCTTGAAGTCTCCCTTG CTTCCATTGAAGGGTCTG 267
EGFR TTAGCAGGAAAGGCACTG CAGCTTCATCCTACACAAG 269
58
PTEN GACAGACTGATGTGTATACG GTGTAAATAGCTGGAGATGG 269
BRAF ATGGTGATGTGGCAGTGAAA TAGCCAGTTGTGGCTTTGTG 150
ESR I CTTGTGCAGGATTGTTGTG GCCAATTGTAGGAACACAG 269 68
NHer 2 AGTACCTGGGTCTGGACGTG CTGGGAACTCAAGCAGGAAG 194
51
N1cRAS AGGGCAGTTTGGATAGCTCA CACCACCCCAAAATCTCAAC 153
Observation in the change of Gene Expression value and its significance
[094] The breast cancer gene expression value profile is characterized by
analyzing the
hereditary genes such as BRCA1 and also the target oncogenes relative gene
expression
value pattern involved in the oncogenesis and metastasis.
_ [095] BRCA1 Gene Expression value Analysis
[096] The breast cancer hereditary gene BRCA I relative gene expression value
in the
patients sample is analyzed using RT-PCR and is compared with the house
keeping gene,
GADPH. The amplification details were provided in the following table:
22-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
Table 29:
Patient Ct Ct Ct Ct
Sample Value Value Sample Value Value Delta
Name 1 2 Mean Name 1 2 Mean Ct
1 BRCA1- GAPDH-
Samp 1 28.01 28.03 28.02 Sampl 25.25 25.28
25.265 2.755
2 BRCA1- GAPDH-
Samp2 26.51 26.18 26.345 Samp2 24.36 24.33 24.345 2
3 BRCA1- GAPDH-
Samp3 26.04 25.18 25.61 Samp3 23.53 23.38 23.455 2.155
4 BRCA1- GAPDH-
Samp4 24.89 25.11 25 Samp4 23.4 23.36 23.38 1.62
Table 30:
delta delta Ct 2 power delta.delta Ct
Patient 1 -0.93 0.524858342
Patient 2 -1.685 0.311002913
Patient 3 -1.53 0.346277367
Patient 4 -2.065 0.238986329
Cancer Gene Specific Analysis
[097] The specific breast cancer genes relative gene expression value in the
patients and
control sample is analyzed using RT-PCR and is compared with the house keeping
gene,
GADPH. The amplification details were provided in the following table:
Table 31:
Ct Ct Ct Ct
Value Value Sample Value Value Delta
Sample Name 1 2 Mean Name 1 2 Mean Ct
GAPDH-
EGFR-Patient 1 33.79 33.67 33.73 Patient 1 24.55 25.03
24.79 8.94
PTEN-Patient 1 29.68 29.23 29.46 4.665
-23-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
BRAF-Patient 1 23.3 24.52 23.91 -0.88
GAPDH-
EGFR-Patient 2 28.68 27.6 28.14 Patient 2 24.27 23.15
23.71 4.43
PTEN-Patient 2 27.1 26.85 26.98 3.265
BRAF-Patient 2 23.33 21.24 22.29 1.425
GAPDH-
EGFR-Patient 3 28.17 26.43 27.3 Patient 3 20.78 20.77
20.78 6.525
PTENPatient 3 26.34 26.41 26.38 5.6
BRAF-Patient 3 22.3 22.74 22.52 1.745
GAPDH-
EGFR-Sample4 27.45 27.98 27.72 Sample4 21.05 20.95 21 6.715
PTENSample4 27.04 27.11 27.08 6.075
BRAFSamp1e4 22.05 22.26 22.16 1.155
Table 32:
Sample Name Ct Value 1 Ct Value 2 Mean Delta Ct
Control-EGFR 32.36 29.11 30.74 9.885
Control-PTEN 25.33 25.4 25.37 4.515
Control-BRAF 21.2 21.47 21.34 0.485
Control-
GAPDH 20.79 20.91 20.85
-24-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
Table 33:
Ct Ct Ct Ct
Sample Value Value Sample Value Value Delta
Name 1 2 Mean Name 1 2 Mean Ct
Patient 3- Patient 3-
Her2 29.54 29.54 29.54
GAPDH 24.6 24.28 24.44 5.1
Patient 3-
IcRAS 21.05 21.34 21.2 3.245
Her2- Sample4-
Patient 4 29.56 29.84 29.7 GAPDH 23.95 30.03 26.99
2.71
lcRAS-
Patient 4 21.86 22.99 22.43 4.565
Table 34:
Sample Name Ct Value 1 Ct Value 2 Mean Delta Ct
Control-Her2 23.28 23.71 23.495 2.35
Control-IcRAS 21.07 21.7 21.385 0.24
Control-
GAPDH 21.13 21.16 21.145
[098] Relative Gene Expression value Analysis: Absolute Relative
quantification
determines the input copy number, usually by relating the PCR signal. Relative

quantification relates the PCR signal of the target transcript in a treatment
group to that of
another sample such as an untreated control. The 2(-Delta Delta C(T)) method
is a
convenient way to analyze the relative changes in gene expression value from
real-time
quantitative PCR experiments.
-25-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Table 35:
delta delta Ct 2 power delta delta Ct
EGFR Patient 1 -0.945 0.52
Patient 2 0.15 1.11
Patient 3 -1.365 0.39
Patient 4 -3.17 0.11
PTEN Patient 1 0.15 1.11
Patient 2 -1.25 0.42
Patient 3 1.085 2.12
Patient 4 1.56 2.95
BRAF Patient 1 -1.365 0.39
Patient 2 -1.91 0.27
Patient 3 1.26 2.39
Patient 4 0.67 1.59
Her2 Patient 3 2.75 6.73
Patient 4 0.36 1.28
kRAS Patient 3 -3.485 0.09
Patient 4 -4.805 0.04

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Gene Expression in Breast Tumor Analysis
[099] A gene expression study was designed and conducted with the primary goal
to
molecularly characterize gene expression in Biopsy or Parafilm embedded tissue
samples
of invasive breast ductal carcinoma, and to explore the correlation between
such
molecular profiles and disease-free survival. Further details of the
embodiment will be
described in the following non-limiting example.
[100] Excise the tissue sample from the animal or remove it from storage.
Remove
RNAlater stabilized tissues from the reagent using forceps. Determine the
amount of
tissue. Do not use more than 30 mg. Weighing tissue is the most accurate way
to
determine the amount. If using the entire tissue, place it directly into a
suitably sized
vessel for disruption and homogenization. If using only a portion of the
tissue, cut it on a
clean surface. Weigh the piece to be used, and place it into a suitably sized
vessel for
disruption and homogenization. Disrupt the tissue and homogenize the lysate in
Buffer
RLT Plus (do not use more than 30 mg tissue).
Table 36: Volumes of Buffer RLT Plus for tissue disruption and homogenization
Amount of starting material Volume of Buffer RLT Plus
<20 mg 350 p.1 or 600 pi
20-30 mg 600 1.t1
[101] Disruption and homogenization using the TissueRuptor: Place the weighed
(fresh,
frozen, or RNAlater stabilized) tissue in a suitably sized vessel. Add the
appropriate
volume of Buffer RLT Plus (see Table 36). Immediately disrupt and homogenize
the
tissue until it is uniformly homogeneous (usually 20-40 s).Centrifuge
thelysate for 3 min
at maximum speed. Carefully remove the supernatant by pipetting, and transfer
it to a
gDNA Eliminator spin column placed in a 2 ml collection tube (supplied).
Centrifuge for
30 s at >8000 x g (>10,000 rpm). Discard the column, and save the flow-
through. This
step is important, as it removes insoluble material that could clog the gDNA
Eliminator
spin column and interfere with DNA removal. In some preparations, very small
amounts
of insoluble material will be present after the 3 mm centrifugation, making
the pellet
invisible. Add 1 volume (usually 350 IA or 600 [1.1) of 70% ethanol to the
flowthrough,
and mix well by pipetting. Do not centrifuge. Proceed immediately to step 6.
If some
-27-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
lysate was lost during homogenization and DNA removal, adjust the volume of
ethanol
accordingly. Transfer up to 700 1 of the sample, including any precipitate
that may have
formed, to an RNeasy spin column placed in a 2 ml collection tube (supplied).
Close the
lid gently, and centrifuge for 15 s at 2:8000 x g 10,000 rpm). Discard the
flow-through.
Add 700 1 Buffer RW1 to the RNeasy spin column. Close the lid gently, and
centrifuge
for 15 s at 28000 x g (2:10,000 rpm) to wash the spin column membrane. Discard
the
flow-through. Add 500 gl Buffer RPE to the RNeasy spin column. Close the lid
gently,
and centrifuge for 15 s at 2:8000 x g (2:10,000 rpm) to wash the spin column
membrane.
Discard the flow-through. Add 500 1 Buffer RPE to the RNeasy spin column.
Close the
lid gently, and centrifuge for 2 min at 2:8000 x g (> 1 0 , 0 0 0 rpm) to wash
the spin column
membrane. Place the RNeasy spin column in a new 1.5 ml collection tube
(supplied). Add
30-50 pl RNase-free water directly to the spin column membrane. Close the lid
gently,
and centrifuge for 1 min at 2:8000 x g (2:10,000 rpm) to elute the RNA. If the
expected
RNA yield is >30 pig, repeat step 11 using another 30-50 1 of RNase-free
water, or using
the eluate from step 11 (if high RNA concentration is required). The rest of
the test steps
are described in previous sections for determining the gene expression value
for a given
portfolio of genes. The results are disclosed below.
Table 37 a, b and c: Her 2 and kRAS Panel: Mean of the 3 Ct values for samples
OT1,
OB1 and 0B3.
Ct
Sample Name 1 Ct 2 Ct 3 Mean
OTI-Her2 29 28.57 28.56 28.68
OTl-IcRAS 28 28.17 28.06 28.09
OB1-Her2 32 32.2 32.1 32.08
OBI-kRAS 29 28.56 28.59 28.66
0B2-Her2 32 31.34 31.25 31.57
0B2-kRAS 28 28.3 28.36 28.28
-28-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
[102] Delta Ct value (Mean of OT1 Her2 minus Mean of OT1 GAPDH). Delta Ct
value
(Mean of OT1 kRAS minus Mean of OT1 GAPDH)
Sample Name Ct 1 Ct 2 Ct 3 Mean Delta Ct
OT1-GAPDH 20.13 20.15 20.04 20.11 8.57
7.99
OBI-GAPDH 19.51 19.44 19.5 19.48 12.60
9.18
0B2-GAPDH 18.7 19.38 19.28 19.12 12.45
9.16
[103] Delta Ct value for Control Her2 (delta Ct Her2 minus Delta Ct GAPDH).
Delta Ct
value for Control Her2 (delta CtkRAS minus Delta Ct GAPDH).
Sample Name Ct 1 Ct 2 Ct 3 Mean Delta Ct
29.1
C12-Her2 29.21 29.06 7 29.15 11.02
25.9
C12-1(RAS 26.19 26.12 1 26.07 7.94
18.2
C12-GAPDH 18.09 18.07 3 18.13
[104] Delta Delta Ct value for OT1 sample (Delta Ct of OT1 Her2 minus Delta Ct
value
of Control).
2 power delta delta. delta
delta delta Ct Ct
-29-

CA 02818890 2013-05-23
WO 2012/070056 PCT/IN2011/000417
OTI-
Her2 -2.45 5.45
OBI-
Her 2 Her2 1.58 0.33
0B2-
Her2 1.44 0.37
OT1-
1cRAS 0.04 Ø97
OBI-
IcRAS 1.24 0.42
=
OB2-
kRAS IcRAS 1.22 0.43
[105] From the above delta delta Ct values, it can be inferred that OT1 sample
is over
expressing Her2 as compared to the control.
Table 38 a, b, c, d for ER:
Sample Name Ct 1 Ct 2 Ct 3 Mean
OTI-ESRI 27.9 27.14 27.05 27.36
OB1-ESR1 31.17 31.24 30.98 31.13
0B3-ESR1 30.91 30.41 30.23 30.52
Sample
Name Ct 1 Ct 2 Ct 3 Mean Delta Ct
OTI-
GAPDH 21.44 21.34 21.28 21.35 6.01
OBI-
GAPDH 20.62 20.53 20.82 20.66 .. 10.47
OB3-
20.55 20.33 20.43 20.44 10.08
-30-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
GAPDH
--31-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
Delta
Sample Name Ct 1 Ct 2 Ct 3 Mean Ct
C12-ESR1 29.62 28.78 29.02 29.14 9.94
C12-GAPDH 19.25 19.17 19.17 19.20
delta delta Ct 2 power delta. delta Ct
OT1-
ER ER -3.93 15.28
OB1-
' ER 0.53 0.69
0B2-
ER 0.14 0.91
[107] From the above Delta Delta Ct values, it can be inferred that sample OTI
is over
expressing ER.
Table 39 a, b, c and d for MET & BRCA2:
Sample Name Ct 1 Ct 2 Ct 3 Mean
OBI-MET 31.51 31.44 31.07 31.34
OB1-BRCA2 30.51 31.02 30.76 30.76
0B3-MET 31.06 30.52 30.82 30.80
0B3-BRCA2 30.25 30.09 33.68 31.34
OT1-MET 27.33 27.26 27.16 27.25
OT1-BRCA2 28.37 28.29 28.44 28.37
-32-

CA 02818890 2013-05-23
WO 2012/070056
PCT/1N2011/000417
Sample Name Ct 1 Ct 2 Ct 3 Mean Delta Ct
OB I -GAPDH 20.82 20.64 20.72 20.73 10.61
10.04
0B3-GAPDH 20.36 20.44 20.19 20.33 10.47
11.01
OT1-GAPDH 21.37 21.23 20.96 21.19 6.06
7.18
Sample Name Ct 1 Ct 2 Ct 3 Mean Delta
C12-MET 30. 30.58 30.26 30.53 11.14
C12-BRCA2 28. 28.81 28.8 28.76 9.37
C12-GAPDH 19. 19.33 19.45 19.39
delta delta Ct 2 power delta ddelta Ct
OB1-MET -0.53 1.44
MET 0B3-MET -0.67 .. 1.59
OTI-MET -5.08 33.82
OBI-
BRCA2 BRCA2 0.67 0.63
1.64 0.32
-2.19 4.56
-33-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
[108] From the above table, MET is being over expressed in the OT1 sample.
Table 39 a, b, c and d for BRCA1, EGFR, PTEN, BRAF:
Sample
Name Ct 1 Ct 2 Ct 3 Mean
OT1-
BRCAI 2951 29.69 30.04 29.75
0T1-EGFR 27.88 28.06 27.93 27.96
OT1-PTEN 28.2 28.3 28.05 28.18
OTI -BRAF 27.33 30.37 28.85
OBI-
BRCAI 31.58 30.67 30.91 31.05
OBI-EGFR 31.91 31.08 31.66 31.55
OBI-PTEN 25.66 25.57 25.4 25.54
OBI-
BRAF 26.62 27.14 26.88
OB3-
BRCA1 30.85 31.35 31.01 31.07
0B3-EGFR 31.74 31.39 31.2 31.44
0B3-PTEN 25.43 25 25.38 25.27
OB3-
BRAF 26.49 28.03 27.26
Sample Name Ct 1 Ct 2 Ct 3 Mean Delta Ct
OT1-GAPDH 21.73 21.53 21.61 21.62 8.12
6.33
6.56
7.23
-34-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
OBI-GAPDH 21.18 21.03 21.2 21.14 9.92
10.41
4.41
5.74
0B3-GAPDH 21.19 21.18 20.93 21.1 9.97
10.34
4.17
6.16
Sample Name Ct 1 Ct 2 Ct 3 Mean Delta Ct
C15-BRCA1 30.1 29.56 30.08 29.91 9.53
C15-EGFR 30.83 30.28 31.02 30.71 10.33
C15-PTEN 23.71 23.9 23.66 23.76 3.38
C15-BRAF 25.06 24.93 25.00 4.62
C15-GAPDH 20.82 20.24 20.08 20.38
delta delta Ct 2 power delta ddelta Ct
OT1 -1.41 2.66
BRCA1 OB1 0.38 0.77
0B3 0.44 0.74
EGFR OT1 -4.00 15.96
OB1 0.08 0.94
0B3 0.01 0.99
-35-

CA 02818890 2013-05-23
WO 2012/070056
PCT/IN2011/000417
PTEN OT1 3.18 0.11
081 1.03 0.49
0B3 0.79 0.58
BRAF OT1 2.61 0.16
OB1 1.13 0.46
0B3 1.55 0.34
[109] From the above table it can be inferred that EGFR is being over
expressed in the
OT 1 sample. All the other genes seem to be normally expressing.
[110] Personalized Treatment: Gene expression value analysis provides various
insights on up regulation or down regulation of multiple genes. In Figure 4,
the upper
horizontal bar 402 represents genes affected by copy number variations (CNV)
or
Mutations and their frequency. The middle bar 404 indicates the target of
potential signal
transduction therapies associated with the particular molecular subtype. The
lower bar
indicates 406 as an example if clinical treatment is done by epidermal growth
factor
receptors (EGFR) targeted by tyrosine kinase (TK) inhibitors. Activation of
downstream
signaling molecules confers resistance to EGFR inhibitors, but may provide an
ideal
target for other therapies. AKT, protein kinase B; CR, complete response;
HER2, human
epidermal growth factor receptor-2; MEK, mitogen-activated protein kinase;
mTOR,
mammalian target of rapamycin; PD, progressive disease; PR, partial response;
PIK3CA,
phosphoinositide 3-kinase; PTEN, phosphatase and tensin homologue; SD, stable
disease.
-36- -

Representative Drawing

Sorry, the representative drawing for patent document number 2818890 was not found.

Administrative Status

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Administrative Status , Maintenance Fee  and Payment History  should be consulted.

Administrative Status

Title Date
Forecasted Issue Date 2022-07-12
(86) PCT Filing Date 2011-06-21
(87) PCT Publication Date 2012-05-31
(85) National Entry 2013-05-23
Examination Requested 2016-03-21
(45) Issued 2022-07-12

Abandonment History

There is no abandonment history.

Maintenance Fee

Last Payment of $125.00 was received on 2023-06-08


 Upcoming maintenance fee amounts

Description Date Amount
Next Payment if small entity fee 2024-06-21 $125.00
Next Payment if standard fee 2024-06-21 $347.00

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Payment History

Fee Type Anniversary Year Due Date Amount Paid Paid Date
Application Fee $200.00 2013-05-23
Maintenance Fee - Application - New Act 2 2013-06-21 $50.00 2013-05-23
Maintenance Fee - Application - New Act 3 2014-06-23 $50.00 2014-06-23
Maintenance Fee - Application - New Act 4 2015-06-22 $50.00 2015-05-15
Request for Examination $400.00 2016-03-21
Maintenance Fee - Application - New Act 5 2016-06-21 $100.00 2016-03-21
Maintenance Fee - Application - New Act 6 2017-06-21 $100.00 2017-02-27
Maintenance Fee - Application - New Act 7 2018-06-21 $100.00 2018-06-20
Expired 2019 - The completion of the application $200.00 2018-11-01
Maintenance Fee - Application - New Act 8 2019-06-21 $100.00 2019-05-23
Maintenance Fee - Application - New Act 9 2020-06-22 $100.00 2020-06-03
Maintenance Fee - Application - New Act 10 2021-06-21 $125.00 2021-06-11
Final Fee 2022-05-04 $152.69 2022-04-29
Maintenance Fee - Application - New Act 11 2022-06-21 $125.00 2022-06-15
Maintenance Fee - Patent - New Act 12 2023-06-21 $125.00 2023-06-08
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
KRISANI BIOSCIENCES (P) LTD
Past Owners on Record
None
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Examiner Requisition 2019-12-31 5 295
Amendment 2020-04-29 8 241
Claims 2020-04-29 2 101
Examiner Requisition 2020-12-10 3 153
Amendment 2021-04-09 7 165
Claims 2021-04-09 2 56
Interview Record with Cover Letter Registered 2021-10-27 1 19
Amendment 2021-11-02 6 109
Claims 2021-11-02 2 56
Final Fee 2022-04-29 5 113
Cover Page 2022-06-14 1 40
Electronic Grant Certificate 2022-07-12 1 2,527
Abstract 2013-05-23 1 65
Claims 2013-05-23 3 91
Drawings 2013-05-23 4 125
Description 2013-05-23 36 1,343
Cover Page 2013-08-16 1 39
Description 2013-05-24 36 1,333
Maintenance Fee Payment 2018-06-20 2 54
Change of Agent 2018-06-20 3 103
Office Letter 2018-06-27 1 23
Office Letter 2018-06-27 1 25
Non-Compliance for PCT - Incomplete 2018-08-21 1 63
Completion Fee - PCT 2018-11-01 3 77
Sequence Listing - New Application / Sequence Listing - Amendment 2018-11-01 3 77
Examiner Requisition 2019-01-28 4 233
Amendment 2019-07-04 9 295
Change to the Method of Correspondence 2019-07-04 3 76
Claims 2019-07-04 2 70
Description 2019-07-04 36 1,393
PCT 2013-05-23 10 428
Assignment 2013-05-23 9 214
Prosecution-Amendment 2013-05-23 3 86
Request for Examination 2016-03-21 1 37

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :